Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Publication year range
1.
FASEB J ; 34(9): 12367-12378, 2020 09.
Article in English | MEDLINE | ID: mdl-32686868

ABSTRACT

HMGB2, a DNA-binding protein, highly expresses during embryogenesis and plays an important role in development of some organs and tissues. However, it remains to be further investigated weather HMGB2 influences muscle development. In this work, we identified HMGB2 as an essential factor in myogenesis. Compared to wild type (WT) mice, body weights of systemic hmgb2 homozygous knockout (hmgb2-/- ) mice especially males were reduced. Diameter and cross-section area of tibialis anterior (TA) muscle fibers as well as expression of Myogenin and MyHC were all decreased in hmgb2-/- mice. CTX injury model revealed that HMGB2 was required for satellite cell proliferation and muscle regeneration. Moreover, HMGB2 interacted with S6K1 and regulated the kinase activity of S6K1 during cell proliferation. Knockdown and inactivation of S6K1 in C2C12 cells both resulted in impaired proliferation and differentiation. Furthermore, expression of cyclin D1 and Myf5 were both decreased when HMGB2 or S6K1 were knocked down and kinase activity of S6K1 was inhibited. These results indicate that HMGB2 is required for skeletal muscle development and regeneration, and HMGB2 maintains proliferation of myoblasts through regulating kinase activity of S6K1.


Subject(s)
HMGB2 Protein/physiology , Muscle Development/physiology , Muscle, Skeletal/physiology , Ribosomal Protein S6 Kinases, 90-kDa/physiology , Animals , Cells, Cultured , Female , Male , Mice , Mice, Inbred C57BL , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/physiology , Regeneration , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/physiology
2.
FASEB J ; 33(12): 14307-14324, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31661640

ABSTRACT

High-mobility group box (HMGB)1 and HMGB2 proteins are the subject of intensive research because of their involvement in DNA replication, repair, transcription, differentiation, proliferation, cell signaling, inflammation, and tumor migration. Using inducible, stably transfected human embryonic stem cells (hESCs) capable of the short hairpin RNA-mediated knockdown (KD) of HMGB1 and HMGB2, we provide evidence that deregulation of HMGB1 or HMGB2 expression in hESCs and their differentiated derivatives (neuroectodermal cells) results in distinct modulation of telomere homeostasis. Whereas HMGB1 enhances telomerase activity, HMGB2 acts as a negative regulator of telomerase activity in the cell. Stimulation of telomerase activity in the HMGB2-deficient cells may be related to activation of the PI3K/protein kinase B/ glycogen synthase kinase-3ß/ß-catenin signaling pathways by HMGB1, augmented TERT/telomerase RNA subunit transcription, and possibly also because of changes in telomeric repeat-containing RNA (TERRA) and TERRA-polyA+ transcription. The impact of HMGB1/2 KD on telomerase transcriptional regulation observed in neuroectodermal cells is partially masked in hESCs by their pluripotent state. Our findings on differential roles of HMGB1 and HMGB2 proteins in regulation of telomerase activity may suggest another possible outcome of HMGB1 targeting in cells, which is currently a promising approach aiming at increasing the anticancer activity of cytotoxic agents.-Kucírek, M., Bagherpoor, A. J., Jaros, J., Hampl, A., Stros, M. HMGB2 is a negative regulator of telomerase activity in human embryonic stem and progenitor cells.


Subject(s)
HMGB2 Protein/physiology , Human Embryonic Stem Cells/enzymology , Stem Cells/enzymology , Telomerase/metabolism , Cell Differentiation , HMGB1 Protein/genetics , HMGB2 Protein/genetics , Human Embryonic Stem Cells/cytology , Humans , Stem Cells/cytology , Transcription, Genetic , Transfection
3.
Hum Cell ; 31(1): 1-9, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28916968

ABSTRACT

The high-mobility group box (HMGB) family includes four members: HMGB1, 2, 3 and 4. HMGB proteins have two functions. In the nucleus, HMGB proteins bind to DNA in a DNA structure-dependent but nucleotide sequence-independent manner to function in chromatin remodeling. Extracellularly, HMGB proteins function as alarmins, which are endogenous molecules released upon tissue damage to activate the immune system. HMGB1 acts as a late mediator of inflammation and contributes to prolonged and sustained systemic inflammation in subjects with rheumatoid arthritis. By contrast, Hmgb2 -/- mice represent a relevant model of aging-related osteoarthritis (OA), which is associated with the suppression of HMGB2 expression in cartilage. Hmgb2 mutant mice not only develop early-onset OA but also exhibit a specific phenotype in the superficial zone (SZ) of articular cartilage. Given the similar expression and activation patterns of HMGB2 and ß-catenin in articular cartilage, the loss of these pathways in the SZ of articular cartilage may lead to altered gene expression, cell death and OA-like pathogenesis. Moreover, HMGB2 regulates chondrocyte hypertrophy by mediating Runt-related transcription factor 2 expression and Wnt signaling. Therefore, one possible mechanism explaining the modulation of lymphoid enhancer binding factor 1 (LEF1)-dependent transactivation by HMGB2 is that a differential interaction between HMGB2 and nuclear factors affects the transcription of genes containing LEF1-responsive elements. The multiple functions of HMGB proteins reveal the complex roles of these proteins as innate and endogenous regulators of inflammation in joints and their cooperative roles in cartilage hypertrophy as well as in the maintenance of joint tissue homeostasis.


Subject(s)
Arthritis, Rheumatoid/genetics , HMGB1 Protein/physiology , HMGB2 Protein/physiology , Osteoarthritis/genetics , Alarmins , Animals , Arthritis, Rheumatoid/immunology , Cartilage, Articular/metabolism , Chondrocytes/pathology , Chromatin Assembly and Disassembly/genetics , Core Binding Factor Alpha 1 Subunit , DNA/metabolism , Disease Models, Animal , Gene Expression , HMGB1 Protein/metabolism , HMGB2 Protein/metabolism , Humans , Hydroxyethylrutoside , Hypertrophy/genetics , Inflammation Mediators , Lymphoid Enhancer-Binding Factor 1 , Mice , Osteoarthritis/immunology , Protein Binding , Wnt Signaling Pathway
5.
Inflammation ; 33(4): 235-43, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20087639

ABSTRACT

Bacterial Lipopolysaccharide (LPS) induced inflammation is implicated in the infection associated testicular tissue damage. Earlier, using a LPS induced acute endotoxemic rat model, we have shown the involvement of inflammation-induced oxidative stress in the impaired steroidogenesis and spermatogenesis. In the present study, we report a significant induction (more than 2-fold) of stress response proteins HSP-60, HMGB-1 and 2 in the testes, as early as 6 h after LPS injection with a later decrease. This induction of acute stress is closely followed by a significant reduction (74%) in Bcl2/Bax ratio along with leakage of cytochrome c (3 fold increase, p < 0.05) from mitochondria and increased caspase-3 activity levels (2.9 fold, p < 0.05) at 12 h and 24 h post LPS injection respectively. Further studies on PARP cleavage revealed a pattern similar to necrotic death during early periods (3 h to 24 h) and apoptosis at later periods (24 h to 72 h) after LPS treatment. In conclusion, the present study shows the involvement of stress response proteins and mitochondrial dysfunction in LPS-induced germ cell death in male rats.


Subject(s)
Chaperonin 60/physiology , Endotoxemia/physiopathology , Endotoxins/toxicity , HMGB1 Protein/physiology , HMGB2 Protein/physiology , Leydig Cells/drug effects , Mitochondria/physiology , Orchitis/chemically induced , Spermatogenesis/drug effects , Acute Disease , Animals , Apoptosis/drug effects , Caspase 3/biosynthesis , Caspase 3/genetics , Chaperonin 60/biosynthesis , Chaperonin 60/genetics , Cytochromes c/metabolism , Endotoxemia/chemically induced , HMGB1 Protein/biosynthesis , HMGB1 Protein/genetics , HMGB2 Protein/biosynthesis , HMGB2 Protein/genetics , Leydig Cells/metabolism , Male , Orchitis/physiopathology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/biosynthesis , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/physiology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Rats , Rats, Wistar , Spermatogenesis/physiology , bcl-2-Associated X Protein/biosynthesis , bcl-2-Associated X Protein/genetics
6.
Blood ; 115(3): 687-95, 2010 Jan 21.
Article in English | MEDLINE | ID: mdl-19965638

ABSTRACT

Gfi-1B is a transcriptional repressor that is crucial for erythroid differentiation: inactivation of the GFI1B gene in mice leads to embryonic death due to failure to produce differentiated red cells. Accordingly, GFI1B expression is tightly regulated during erythropoiesis, but the mechanisms involved in such regulation remain partially understood. We here identify HMGB2, a high-mobility group HMG protein, as a key regulator of GFI1B transcription. HMGB2 binds to the GFI1B promoter in vivo and up-regulates its trans-activation most likely by enhancing the binding of Oct-1 and, to a lesser extent, of GATA-1 and NF-Y to the GFI1B promoter. HMGB2 expression increases during erythroid differentiation concomitantly to the increase of GfI1B transcription. Importantly, knockdown of HMGB2 in immature hematopoietic progenitor cells leads to decreased Gfi-1B expression and impairs their erythroid differentiation. We propose that HMGB2 potentiates GATA-1-dependent transcription of GFI1B by Oct-1 and thereby controls erythroid differentiation.


Subject(s)
Cell Differentiation/genetics , Erythrocytes/physiology , HMGB2 Protein/physiology , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Base Sequence , Cell Differentiation/drug effects , Cells, Cultured , Erythrocytes/drug effects , Erythrocytes/metabolism , Erythroid Cells/drug effects , Erythroid Cells/metabolism , GATA1 Transcription Factor/metabolism , GATA1 Transcription Factor/physiology , HMGB2 Protein/antagonists & inhibitors , HMGB2 Protein/genetics , HMGB2 Protein/metabolism , HeLa Cells , Humans , Molecular Sequence Data , Octamer Transcription Factor-1/metabolism , Octamer Transcription Factor-1/physiology , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins/metabolism , RNA, Small Interfering/pharmacology , Repressor Proteins/metabolism , Transcriptional Activation/drug effects
7.
Cancer Invest ; 26(8): 843-51, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18798064

ABSTRACT

High mobility group B (HMGB) chromosomal proteins, which plays important role in cancer and inflammation, were followed at various stages of the squamous cell carcinoma of skin. Present results were analyzed by histopathology, BrdU assay, immunohistochemistry, western blot and RT-PCR, which indicate that at early stages of tumorigenesis, expression of HMGB (B1, B2), raised only by about 20%. However, the advanced (> or =12 weeks) tumors showed significant (> or =80%) increase in HMG levels. Using skin cancer model, we demonstrated that high levels of HMGB directly correlate with the extent of neoplastic changes, and it appears that HMGB is an effective stimulus for cell differentiation, tumor progression, and metastatic invasion.


Subject(s)
Carcinoma, Squamous Cell/chemistry , HMGB1 Protein/analysis , HMGB2 Protein/analysis , Neoplasm Proteins/analysis , Skin Neoplasms/chemistry , Animals , Benzo(a)pyrene , Carcinoma, Squamous Cell/chemically induced , Carcinoma, Squamous Cell/pathology , Cell Differentiation , DNA Replication , Disease Progression , Female , HMGB1 Protein/physiology , HMGB2 Protein/physiology , Mice , Neoplasm Invasiveness , Neoplasm Proteins/physiology , Skin Neoplasms/chemically induced , Skin Neoplasms/pathology
8.
Nat Cell Biol ; 9(4): 402-14, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17384639

ABSTRACT

Nuclear dysfunction is a key feature of the pathology of polyglutamine (polyQ) diseases. It has been suggested that mutant polyQ proteins impair functions of nuclear factors by interacting with them directly in the nucleus. However, a systematic analysis of quantitative changes in soluble nuclear proteins in neurons expressing mutant polyQ proteins has not been performed. Here, we perform a proteome analysis of soluble nuclear proteins prepared from neurons expressing huntingtin (Htt) or ataxin-1 (AT1) protein, and show that mutant AT1 and Htt similarly reduce the concentration of soluble high mobility group B1/2 (HMGB1/2) proteins. Immunoprecipitation and pulldown assays indicate that HMGBs interact with mutant AT1 and Htt. Immunohistochemistry showed that these proteins were reduced in the nuclear region outside of inclusion bodies in affected neurons. Compensatory expression of HMGBs ameliorated polyQ-induced pathology in primary neurons and in Drosophila polyQ models. Furthermore, HMGBs repressed genotoxic stress signals induced by mutant Htt or transcriptional repression. Thus, HMGBs may be critical regulators of polyQ disease pathology and could be targets for therapy development.


Subject(s)
HMGB1 Protein/physiology , HMGB2 Protein/physiology , Neurodegenerative Diseases/metabolism , Nuclear Proteins/physiology , Proteomics/methods , Animals , Blotting, Western , Cell Death , Cells, Cultured , Drosophila , Electrophoresis, Gel, Two-Dimensional , HMGB1 Protein/analysis , HMGB1 Protein/metabolism , HMGB2 Protein/analysis , HMGB2 Protein/metabolism , Immunohistochemistry , Immunoprecipitation , Models, Biological , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Neurons/cytology , Neurons/metabolism , Nuclear Proteins/analysis , Nuclear Proteins/metabolism , Peptides/genetics , Peptides/metabolism , Protein Binding , Purkinje Cells/cytology , Purkinje Cells/metabolism , RNA, Small Interfering , Rats , Rats, Wistar , Signal Transduction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
9.
DNA Cell Biol ; 21(3): 199-212, 2002 Mar.
Article in English | MEDLINE | ID: mdl-12015897

ABSTRACT

Overcoming local DNA rigidity is required to perform three-dimensional DNA-protein configuration at promoter regions. The abundant architectural nonhistone chromosomal HMG box proteins are nonsequence-specific; however, they have been established to specifically recognize distorted DNA. Using transient transfection to overexpress two different members of the HMGB-1/2 family of DNA architectural factors, we demonstrate that these proteins provide a general enhancement in reporter gene expression irrespective of the promoter being considered. Evidences are also provided indicating that stimulation may not be achieved by recruitment of the proteins by regulatory factors or as a consequence of major chromatin unfolding as previously suggested. Interestingly, the influence of the HMG box proteins under study was overridden when the promoters were either induced or stimulated by Trichostatin A (TSA) but recovered upon extended induction period. These results also support the concept that the architectural role of these proteins can contribute to the preinitiation complex assembly required for basal transcription, but to a much lesser extent to the poised promoter scaffolding characteristic of activated transcription.


Subject(s)
HMGB Proteins/physiology , Transcriptional Activation , Cell Line , Chromosomes/chemistry , Genes, Reporter , HMGB Proteins/antagonists & inhibitors , HMGB1 Protein/physiology , HMGB2 Protein/physiology , Humans , Hydroxamic Acids/pharmacology , Kinetics , Promoter Regions, Genetic , Trans-Activators/physiology , Transcriptional Activation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...